bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1 Analysis of contributory gut microbiota and lauric acid against necrotic enteritis in

2 Clostridium perfringens and Eimeria side-by-side challenge model

3

4

5 Short title: The impact of Clostridium perfringens challenge on gut microbiota in chickens

6

7 Wen-Yuan Yang1, Yuejia Lee1, Hsinyi Lu1, Chung-Hsi Chou2, Chinling Wang1*

8

9 1 Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University,

10 P.O. Box 6100, Mississippi State, MS 39762, 2 Zoonoses Research Center and School of

11 Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei City,

12 106, Taiwan

13

14 *Corresponding author: [email protected]

1 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

16 Abstract

17 Gut microbiota has been demonstrated to be involved in intestinal nutrition, defense, and

18 immunity, as well as participating in disease progression. This study was to investigate gut

19 microbiota changes in chickens challenged with netB-positive Clostridium perfringens strain 1

20 (CP1) and/or the predisposing Eimeria (Eimeria). In addition, the effects of lauric acid, a

21 medium-chain fatty acid (MCFA), on NE reduction and modulation of microbiota were

22 evaluated. The results demonstrated that microbial communities in the jejunum were distinct

23 from those in the cecum, and the microbial community change was more significant in jejunum.

24 Challenge of CP1 in conjunction with Eimeria significantly reduced species diversity in jejunal

25 microbiota, but cecal microbiota remained stable. In the jejunum, CP1 challenge increased the

26 abundance of the genera of Clostridium sensu stricto 1, Escherichia Shigella, and Weissella, but

27 significantly decreased the population of Lactobacillus. Eimeria infection on its own was unable

28 to promote NE, demonstrating decrements of Clostridium sensu stricto 1 and Lactobacillus. Co-

29 infection with CP1 and Eimeria reproduced the majority of NE lesions with significant

30 increment of Clostridium sensu stricto 1 and reduction in Lactobacillus. The changes of these

31 two taxa increased the severity of NE lesions. Further analyses of metagenomeSeq, STAMP, and

32 LEfSe showed significant overgrowth of Clostridium sensu stricto 1 was associated with NE and

33 Eimeria infection than C. perfringens challenge alone. The supplementation of lauric acid did

34 not reduce NE incidence and severity but decreased the relative abundance of Escherichia

35 Shigella. In conclusion, significant overgrowth of Clostridium sensu stricto 1 in the jejunm is the

36 major microbiota contributory to NE. Controlling proliferation of this taxon in the jejunum

37 should be the niche for developing effective strategies against NE.

2 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

38 Keywords: necrotic enteritis, Clostridium perfringens, Eimeria, lauric acid, microbiota

3 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

39 Introduction

40 Necrotic enteritis (NE) as the result of proliferations of Clostridium perfringens (C. perfringens)

41 type A and their associated toxins in the small intestine of chickens is a devastating enteric

42 disease, characterized by sudden diarrhea, unexpected mortality, and mucosal necrosis [1, 2]. Up

43 to 37% of commercial broiler flocks is estimated to be affected by this disease, and it has

44 contributed to the losses of 6 billion dollars in the global poultry industry [3, 4]. In recent

45 decades, C. perfringens-associated NE in poultry has been well-controlled by in-feed

46 antimicrobial growth promoters (AGPs) [5]. However, the emergence of antibiotic-resistant

47 from animals and the potential threat of transmission to humans has led to bans on using

48 AGPs in many countries [6, 7]. Following the withdrawal of AGPs from poultry feed, NE has re-

49 emerged as a significant disease to the poultry industry [8-11].

50 Gut microbiota is one of the central defense components in the gastrointestinal tract against

51 enteric pathogens, which works by modulating host responses to limit the colonization of

52 pathogens [12]. Interactions between gut microbiota and the host could influence intestinal

53 morphology, physiology, and immunity [13]. Recently, gut microbiota has been demonstrated to

54 regulate intestinal gene expression [14] and T cell-mediated immunity [15] as well as to

55 accelerate the maturation of the gut immune system [16]. Conversely, a growing number of

56 studies have observed gut microbial shifts in enteric diseases, considering that gut microbiota

57 plays a role in the progress of disease development. Similar results were also represented in NE

58 induction models, proposing that the disturbance of gut microbiota interacts with the host,

59 subsequently promoting the development of NE [17-20]. In the case of human necrotizing

60 enterocolitis, an enteric disease in infants associated with C. perfringens [21], a recent study

61 found that Bacteroides dorei, an opportunist pathogenic bacterium in anaerobic infections, was

4 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

62 associated with an increased mortality of this disease [22]. Furthermore, several studies have

63 demonstrated that the increment of bacteria belong to a genus of Escherichia-Shigella was

64 associated with C. perfringens infection [5, 20]. This evidence raised the possibility that certain

65 microbes or microbiota in the gut may contribute to the virulence or development of enteric

66 disease in chickens, particularly for NE.

67 The removal of AGPs drove the poultry industry to search for an alternative in prevention to

68 decrease the incidence of NE. Probiotics, prebiotics, organic acids, plant extracts, essential oils,

69 and enzymes arose in response to this demand, but the efficacy of those on NE reduction were

70 variable and inconsistent [23, 24]. However, a medium-chain fatty acid (MCFA), lauric acid, was

71 found to have strong in vitro antimicrobial activity against gram-positive organisms [25-27] and

72 C. perfringens [28, 29]. In an in vivo trial, lauric acid with butyric acid demonstrated the lowest

73 incidence and severity of NE compared to other treatments [29]. However, this promising result

74 did not promote more applications of lauric acid against NE, and the interaction of lauric acid

75 with gut microbiota was not even addressed. The evaluation of its modulation effect on NE

76 reduction and gut microbiota simultaneously would be valuable in exploring specific microbial

77 community contributory to NE.

78 Although C. perfringens is the causative etiological agent of NE, it is evident that other

79 predisposing factors are required for NE induction [10, 30-32]. Even though gut microbiota has

80 been suggested to be involved in the progress of NE development [33, 34], association between

81 microbiota profile and NE development have not been well elucidated. Most studies intensively

82 focused on changes of microbial communities in the ileum or cecum where higher quantity of

83 microbes or/and more diverse microbial compositions were harbored; however, most results

84 were inconclusive [17-19, 35, 36]. Inversely, microbiota in the jejunum, which serve as the

5 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

85 primary site for colonization of C. perfringens and development of NE [33], was seldom

86 evaluated. In the present study, we investigated gut microbiota targeting NE cases and in

87 chickens with side-by-side treatments with the causative pathogen and parasitic predisposing

88 factor, Eimeria, and expected to unveil the contributory microbe or microbiota to NE. The

89 effects of lauric acid on NE reduction and modulation of microbiota were also examined to

90 confer the alternative intervention to prevent and control NE.

91

92 Materials and methods

93 Ethics statement

94 All procedures for the care, housing and treatment of chickens were approved by the Institutional

95 Animal Care and Use Committee at Mississippi State University (IACUC 16-439).

96

97 Chicken, diet, and experimental design

98 A total of 50 male and female one-day-old unvaccinated broiler chicks (Cobb strain) were

99 obtained from a commercial hatchery. Chicks were inspected on receiving to ensure their healthy

100 status and randomly allotted to 5 groups, studying the NE incidence and gut microbiota after

101 challenge of netB-positive C. perfringens (A group: CP1), co-infection with netB-positive C.

102 perfringens and multi-species Eimeria (B group: CP1+Eimeria), addition of lauric acid to feed

103 chickens co-infected with netB-positive C. perfringens and multi-species Eimeria (C group:

104 CP1+Eimeria+LA), inoculation of multi-species Eimeria (D group: Eimeria), and no treatment

105 (E group: CTL).

106 Chicks in groups were placed in separate temperature-controlled iron tanks with nets in the

107 floor-pen facility and lined with fresh litter. Throughout the 19-day study period, wheat-based

6 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

108 diets prepared based on the formula by Branton et al. [37] were offered for the first 7 days, and

109 then the rations were replaced by fishmeal diets (wheat-based diets containing 50% fishmeal)

110 obtained from the 1:1 mixture of wheat-based diet with fishmeal 60 N (Seven Springs Farm,

111 Check, Virginia, USA), containing minimal 60% crude protein from days 8 until the end of the

112 study. For the lauric acid supplementing group, 400 mg of lauric acid powder (Fisher Scientific,

113 Pittsburgh, Pennsylvania, USA) was added into 1 kg of wheat-based diet or fishmeal diet to form

114 the final ration for chickens from day 8 onward [29].

115 Co-infection with netB-positive C. perfringens (CP1) and multi-species Eimeria was applied

116 to induce NE according to our previous studies. The success of reproducing NE was determined

117 by clinical signs and intestinal lesion scores reaching 2 or more. In brief, chickens in the co-

118 infection group were given a single gavage of coccidial inoculum at day 10, followed by oral

119 administration of 3 ml CP1 inoculum with average 2.5x108 colony-forming units (CFU)/ml at

120 day 15 for 4 consecutive days with a frequency of 3 times daily. For a single challenge of CP1 or

121 Eimeria, the same methodology and time points were conducted as in the co-infection group. All

122 chickens were inspected on a daily basis and humanely euthanized at day 19 by carbon dioxide.

123 Dead chickens not resulting from NE were excluded from the trial after necropsy. The

124 experimental trial was reviewed and approved by the Mississippi State University Institutional

125 Animal Care and Use Committee.

126

127 Challenge strain and inoculum preparation

128 Anticoccidial live vaccine containing live oocysts of E. acervulina, E. maxima, E. maxima MFP,

129 E. mivati, and E. tenella was used as a disposing factor. .The vaccine bottle contained 10,000

130 doses of oocysts in an unspecified proportion of Eimeria species. A ten-fold dose of vaccine was

7 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

131 prepared then applied on Eimeria-treated and co-infection groups. C. perfringens, a clinical NE

132 strain designated as CP1 obtained from Dr. John F. Prescott (Ontario Agricultural College,

133 University of Guelph, Canada), was used to challenge chickens. This strain was characterized as

134 netB-positive Type A and used to reproduce NE in a number of experiments [38-41]. CP1 was

135 cultured on blood agar plates and incubated anaerobically at 37°C for overnight. A single colony

136 was in turn transferred into 3 ml of fluid thioglycollate (FTG) medium (Himedia, Mumbai,

137 Maharashtra, India) at 37°C for overnight. Thereafter, the bacterial suspension was inoculated

138 into fresh FTG broth at a ratio of 1:10 and incubated at 37°C for 15, 19, and 23 hours,

139 respectively. The whole broth cultures were used to induce NE based on the evidence that

140 with toxins produce more severe disease than using cells alone [38]. The bacterial

141 concentration (CFU/ml) of inoculum was calculated by plate counting using Brain Heart Infusion

142 agar (Sigma-Aldrich, St. Louis, Missouri, USA), followed by anaerobic incubation at 37°C for

143 16 hours.

144

145 Sample collection and lesion scoring

146 Three chickens per group were randomly selected to collect fecal contents from the jejunum (AJ,

147 BJ, CJ, DJ, and EJ) and cecum (AC, BC, CC, DC, and EC). Among three CP1-challenged groups

148 (A, B, and C), chickens suffering NE (lesion score ≥ 2) were preferentially collected. Then, the

149 remaining chickens were sampled randomly to reach a quantity of 3. One percent of 2-

150 mercaptoethanol (Sigma-Aldrich) in PBS was used to wash fecal contents, and samples were

151 immediately frozen at -80°C. The intestinal tissues (duodenum to ileum) were inspected for NE

152 lesions and scored following the criteria described by Keyburn [42], with a range of 0 (no gross

153 lesions), 1 (congested intestinal mucosa), 2 (small focal necrosis or ulceration; one to five foci),

8 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

154 3 (focal necrosis or ulceration; 6 to 15 foci), and 4 (focal necrosis or ulceration; 16 or more foci).

155 Chickens with lesion scores reaching 2 or higher were identified as NE cases, and the highest

156 score in their small intestinal sections (duodenum, jejunum, and ileum) was recorded as the final

157 score of NE.

158

159 DNA extraction

160 Total genomic DNA was isolated from approximately 250 mg of fecal contents using the

161 MOBIO PowerFecal® DNA Isolation Kit (Mobio, Germantown, Maryland, USA) following the

162 manufacturer’s protocol with some modifications. After adding bead solution and lysis buffer,

163 the mixture was heated in a water bath at 65°C for 30 minutes followed by 5 minutes of

164 vortexing. The concentration and quality of harvested DNA were determined by NanoDrop™

165 One Microvolume UV-Vis Spectrophotometer (Fisher Scientific) and visualized on 0.8%

166 agarose gel (BD Biosciences, San Jose, California, USA). Afterward, genomic DNA was stored

167 at -20°C until further analysis.

168

169 16S rRNA library preparation and sequencing

170 The variable V3-V4 region of the 16S rRNA gene was PCR-amplified in 25-μl reaction mixtures,

171 containing 12.5 μl Clontech Labs 3P CLONEAMP HIFI PCR PREMIX (Fisher Scientific), 1 μl

172 of each 10-μm Illumina primer (forward primer-5’CCTACGGGNGGCWGCAG 3’ and reverse

173 primer-5’ GACTACHVGGGTATCTAATCC 3’) with standard adapter sequences, and 1 μl of

174 DNA template. The PCR conditions started with an initial denaturation step at 95°C for 3

175 minutes, followed by 25 cycles of 95°C for 30 seconds, 55°C for 30 seconds, and 72°C for 30

176 seconds, and a final extension step at 72°C for 5 minutes on Applied Biosystems GeneAmp PCR

9 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

177 System 9700 (Applied Biosystems Inc., Foster City, California, USA). The amplicons were

178 cleaned up by Monarch® DNA Gel Extraction Kit (New England Biolabs, Ipswich,

179 Massachusetts, USA). Subsequently, an index PCR was performed by using Nextera XT Index

180 Kit (Illumina, San Diego, California, USA) to attach a unique 8-bp barcode sequence to the

181 adapters. The applied 25-μl reaction was composed of 12.5 μl KAPA HiFi HotStart Ready Mix

182 (Kapa Biosystems, Wilmington, Massachusetts, USA), 2.5μl of each index primer, and 1 μl of

183 16S rRNA amplicon and reaction conditions were as follows: 95°C for 3 minutes, 8 cycles of

184 95°C for 30 seconds, 55°C for 30 seconds, 72°C for 30 seconds, and 72°C for 5 minutes on

185 Mastercycler® pro (Eppendorf AG, Hamburg, Germany). The PCR products were purified using

186 Agencourt AMPure XP beads (Beckman Coulter, Indianapolis, Indiana, USA), and the size and

187 concentration were determined by Bioanalyzer with DNA 1000 chip (Agilent, Santa Clara,

188 California, USA) and Qubit® 2.0 Fluorometer with Qubit™ dsDNA HS Assay Kit (Fisher

189 Scientific). Those libraries were normalized and pooled to one tube with the final concentration

190 of 10 pM. Samples were thereafter sequenced on the MiSeq® System using Illumina MiSeq

191 Reagent Kit v3 (2×300 bp paired-end run).

192

193 Sequence processing and data analysis

194 Paired-end sequences were merged by means of fast length adjustment of short reads (FLASH)

195 v1.2.11 [43] after trimming of primer and adapter sequences. Reads were de-multiplexed and

196 filtered by Quantitative Insights into Microbial Ecology (Qiime) software v1.9.1 [44], meeting

197 the default quality criteria and a threshold phred quality score of Q ≥ 20. Chimeric sequences

198 were filtered out using the UCHIME algorithm [45]. The pick-up of operational taxonomic units

199 (OTUs) was performed at 97% similarity by the UPARSE algorithm [46] in USEARCH [47].

10 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

200 The OTUs were further subjected to the -based analysis by RDP Classifier v2.11 with

201 a cut-off of 80% [48] using the Silva v128 database. Differential abundance of OTU among

202 treatments was evaluated by metagenomeSeq. The clustered OTUs and taxa information were

203 used for diversity and statistical analyses by Qiime v1.9.1 and R package v.3.3.1 (http://www.R-

204 project.org/). Differences of taxonomic profiles between groups were compared using Statistical

205 Analysis Metagenomic Profiles (STAMP) software [49] v2.1.3 with Welch’s t-test.

206 Furthermore, LEfSe (linear discriminant analysis effect size) from the LEfSe tool

207 (http://huttenhower.sph.harvard.edu/lefse/), an algorithm for high-dimensional class comparisons

208 between biological conditions, was used to determine the significant feature taxa between groups

209 or intestinal location. It emphasizes statistical significance, biological consistency, and effect

210 relevance and allows researchers to identify differentially abundant features that are also

211 consistent with biologically meaningful categories [50]. The Kruskal-Wallis rank sum test was

212 included in LEfSe analysis to detect significantly different abundances and performed LDA

213 scores to estimate the effect size (threshold: ≥ 4).

11 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

214 Results

215 NE reproduction and effects of lauric acid as an alternative prevention

216 Six of the NE cases were identified in three CP1-challenged groups (Table 1). They showed

217 different degrees of characteristic gross lesions in small intestinal tissues. The most severe

218 lesions were found in the jejunum, between its proximal end and Meckel’s diverticulum. Under

219 co-infection with CP1 and Eimeria, the incidence and severity of NE increased. No NE mortality

220 was noticed. Statistically significant differences of lesion score (LS) were determined between

221 three CP1-challenged groups (A, B, and C) and the control counterpart (p ≤ 0.05). The co-

222 infection groups (B and C) demonstrated a highly significant difference (p ≤ 0.01). However, the

223 supplementation of lauric acid did not reduce the incidence and severity which were similar to

224 the NE positive control group.

225

226 Table 1. NE frequency and mean lesion score by groups NE lesion score NE Group Treatment Subtotal Lesion score 0 1 2 3 4 case A CP1 0 9 1 0 0 10 1.11 ± 0.31a 1 B CP1+Eimeria 0 8 0 1 1 10 1.50 ± 1.02a 2 C CP1+Eimeria+LA 0 7 1 1 1 10 1.60 ± 1.02a 3 D Eimeria - - - - - 10 - 0 E CTL 5 4 0 0 0 9 0.44 ± 0.50b 0

227 LA lauric acid; NE necrotic enteritis; CTL: control group. 228 NE case: lesion score reaching 2 or above. 229 One chick in CTL was misclassified during the trial and excluded. 230 Dissimilar letters indicate a significant difference at a level of α=0.05.

231

232 Metadata and sequencing

12 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

233 A total of 11,191,102 sequence reads with an average length of 453 ±5 base pairs were obtained

234 from 30 samples, including 15 jejunal samples (3 samples per group in AJ, BJ, CJ, DJ, and EJ)

235 and 15 cecal samples (3 samples per group in AC, BC, CC, DC, and EC). The sequences were

236 filtered and further clustered into OTU using a cut-off of 97% similarity. The estimate of Good’s

237 coverage reached 98% for all the jejunal and cecal samples. The rarefaction curve demonstrated

238 that the sequencing depth was adequate to cover the bacterial diversity in the jejunal and cecal

239 samples (Figure S1).

240

13 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

241 Normal microbial composition in the jejunum and cecum

242 (92.1% of relative abundance) was the most dominant phylum in the jejunum,

243 followed by Cyanobacteria (2.2%) and Proteobacteria (2.1%), Bacteroidetes (1.9%), and

244 Actinobacteria (1.7%). On the contrary, the phylum of Bacteroidetes (75.5%) predominated in

245 the cecum, followed by Firmicutes (19.8%) and Proteobacteria (4.7%) (Figure 1A). At the

246 genus level, jejunal contents were dominated by Lactobacillus (41.2% of relative abundance) and

247 Clostridium sensu stricto 1 (39.1%), followed by other unclassified genus (8.7%), Weissella

248 (3.6%), Enterococcus (1.9%), Escherichia Shigella (1.8%), and Staphylococcus (1.6%).

249 Bacteroides (75.5%) was the most abundant genus in the cecum, followed by other unclassified

250 genus (17.2%), Escherichia Shigella (3.1%), Eisenbergiella (1.7%), and Anaerotruncus (1.5%)

251 (Figure 1B). The genera of Lactobacillus, Clostridium sensu stricto 1, Weissella, Enterococcus,

252 Staphylococcus, and Bifidobacterium in the jejunum exhibited significant difference in

253 abundance compared to those in the cecum. Cecal microbiota contained significantly higher

254 abundance of Bacteroides and Proteus (Welch’s t test, p < 0.05; Figure S2).

255 256

14 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

257 A)

258 259

15 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

260 (B)

261 262

16 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

263 (C)

264 Figure 1. Microbiota composition in jejunum and cecum with different treatments. Each bar

265 represents the average relative abundance of each bacterial taxon within a group. The top 5 and

266 10 abundant taxa are shown at the level of phylum and genus, respectively. (A) Abundant phyla

267 in jejunum and ceca by groups; (B) Abundant genera in jejunum and ceca by groups; (C)

268 Abundant genera in jejunal and cecal samples. LS stands for lesion score of NE.

17 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

269 Changes of microbial communities in response to treatments

270 In the jejunum, challenge of CP1 increased the relative abundance of the genera of Clostridium

271 sensu stricto 1 (54.75%), Escherichia Shigella (9.57%), and Weissella (4.99%) but significantly

272 decreased the population of Lactobacillus (25.44%) (Figure 2). The inoculation of Eimeria to

273 chickens significantly increased the relative abundance of Weissella (16.01%) and

274 Staphylococcus (6.51%), but decreased the amount of Lactobacillus (30.66%) and Clostridium

275 sensu stricto 1 (27.69%). Co-infection with CP1 and Eimeria led to significant increment of

276 Clostridium sensu stricto 1 (71.89%), increased relative abundance of Escherichia Shigella

277 (4.68%), but the decrements of Lactobacillus (16.99%), Weissella (0.44%) and Staphylococcus

278 (0.40%). In the cecum, different treatments did not promote significant difference of taxa

279 abundance between groups with an exception of Eisenbergiella, significantly increased in co-

280 infection group. However, challenge of CP1 and co-infection of CP1 and Eimeria still promoted

281 cecal increments of Clostridium sensu stricto 1 (the relative abundance of this taxon in groups

282 challenging with CP1, CP1 and Eimeria, and control was 0.75%, 1.99%, and 0.02%).

283

284

18 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

285 (A)

286 287 (B)

288

19 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

289 (C)

290 291

20 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

292 (D)

293 294

21 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

295 (E)

296 297

22 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

298 (F)

299 300 Figure 2. Differential abundance of genera between groups by metagenomeSeq. (A)

301 Lactobacillus; (B) Clostridium sensu stricto 1; (C) Weissella; (D) Staphylococcus; (E)

302 Escherichia Shigella; (F) Eisenbergiella. * p ≤ 0.05 and ** p ≤ 0.01.

303

304 Microbial diversities in response to treatments

305 In jejunal microbiota, challenge of CP1 (AJ) and co-infection with CP1 and Eimeria (BJ)

306 reduced species richness and evenness, but the infection of Eimeria (DJ) exerted counter results.

307 Addition of lauric acid into co-infection group (CJ) exacerbated the reduction observed in BJ

308 group. However, no apparent effect was noted on cecal microbiota following above treatments

309 (Figure S3 and Figure 3). Analysis of alpha diversity by Shannon index further demonstrated

310 that challenge of CP1 in conjunction with Eimeria infection significantly reduced species

311 diversity in jejunal microbiota. The 16S rRNA gene survey by principal coordinate analysis

23 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

312 (PCoA) and principal component analysis (PCA) showed a distinct separation of two community

313 profiles between the jejunal and cecal microbiota. Cluster and heat map analyses exhibited

314 distinct classifications and microbial compositions between the jejunum and cecum, coinciding

315 with observations on PCoA and PCA (Figure 4). Additionally, the results of PCoA and PCA

316 also depicted the differential diversity between the CP1-challenged (group AJ, BJ and CJ),

317 Eimeria-infected (DJ), and control (EJ) groups in jejunal microbiota, showing that challenge of

318 CP1 shared similar microbial community structures with co-infection with CP1 and Eimeria.

319 However, cecal groups with CP1 treatments did not display cluster phenomenon as jejunal

320 groups displayed in PCoA. PCA with hierarchical clustering further reflected that Clostridium

321 sensu stricto 1 was contributory to the similarity of NE assemblage, and the genera of

322 Lactobacillus, Weissella, and Staphylococcus contributed to discrepant community structures in

323 Eimeria-treated and control groups in the jejunum. On the other hand, Bacteroidetes was the

324 main genus contributing to the distinct separation between jejunal and cecal groups (Figure 5).

325

326

24 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

327 (A)

328 329

25 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

330 (B)

331 332

26 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

333 (C)

334 335

27 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

336 (D)

337 338 Figure 3. Comparison of microbial diversity between groups in jejunum and cecum using

339 different measures of alpha diversity. (A) Shannon index, (B) Simpson index (C), abundance-

340 based coverage estimator (ACE) index, and (D) Chao1 index. Results are shown as mean ±

341 SEM. Kruskal-Wallis test: * p ≤ 0.05, ** p ≤ 0.01, and *** p ≤ 0.001.

342

28 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

343 (A)

344 345 (B)

346 347

29 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

348 (C)

349 350 (D)

351 352 Figure 4. Comparison of the compositions and similarities of jejunal and cecal microbiota with

353 different treatments. (A) Weighted Unifrac principal coordinate analysis (PCoA); (B) principal

354 component analysis (PCA); (C) cluster analysis by unweighted paired-group method using

30 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

355 arithmetic means (UPGMA) using unweighted Unifrac distance; (D) heat map analysis at the

356 genus level.

357

358 359 Figure 5. Principal component analysis and hierarchical clustering of contributory genus to NE

360 assemblage (in red circle) and to the dissimilarity between groups.

361

362 Microbial community structure and taxa contributory to NE

363 Analysis of jejunal microbiota in NE cases revealed that Clostridium sensu stricto 1, to which

364 causative C. perfringens belongs, was the most dominant genus, followed by Lactobacillus,

365 Weissella, Escherichia Shigella, Staphylococcus, and others. Accompanying the elevation of NE

366 severity, the relative abundance of Clostridium sensu stricto 1 increased (relative abundance ≥

367 75% in LS4 compared to 50-75% in LS2 and LS3). Conversely, the population of Lactobacillus

368 decreased while the lesion score was elevated. The relative amount of Escherichia Shigella was

31 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

369 variable in NE cases, presenting higher abundance after CP1 challenge but low population

370 following co-infection with CP1 and Eimeria. (Figure 1C).

371 Heat map analysis exhibited that NE cases harbored the similar microbial community profile.

372 Clostridium sensu stricto 1 and C. perfringens were consistently presented and abundant taxa in

373 jejunum (Figure 6). Opposite low abundance of Lactobacillus was noted. However, only the

374 increment of Clostridium sensu stricto 1 but not C. perfringens (data not shown) demonstrated

375 significance on NE by metagenomeSeq (Figure 2B). Using Welch's t-test, jejunal groups further

376 showed that CP1 in conjunction with Eimeria increased significantly Clostridium sensu stricto 1

377 and C. perfringens when compared to the control (Figure S4; p < 0.05), whereas challenge of

378 CP1 alone did not lead to significant increase of these taxa. Differential abundant phylotypes

379 between different treatments in jejunum were further evaluated by LEfSe using the LDA score of

380 4. This threshold guarantees that the meaningful taxa is compared and eliminates most of rare

381 taxa. LEfSe demonstrated similar results as Welch’s test that challenge of CP1 unable to yield a

382 significantly higher amount of Clostridium sensu stricto 1 and C. perfringens. However,

383 significant differences were displayed when CP1 co-infected with Eimeria (Figure 7). No

384 differential taxon was found in cecal groups (AC, BC, CC, HDC, and EC) while Welch's t-test

385 and LEfSe were applied.

386

387

32 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

388 (A)

389 390

33 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

391 (B)

392 393

34 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

394 (C)

395

396 397 Figure 6. Heat map analysis of contributory taxa to NE at the genus level in jejunal (A) and

398 cecal (B) samples. (C) Heat map of gut bacteria with the relative abundance of OTUS by z score

35 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

399 and represented bacterial taxa information, including phylum, family, genus, and species. Top 26

400 taxa was shown.

401

402 (A)

403 404

36 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

405 (B)

406 407 Figure 7. LEfSe identified the most differentially abundant clades at all taxonomic levels

408 between jejunal groups using the LDA score of 4. Differentially abundant taxa in group BJ

409 versus EJ (A) and CJ versus EJ (B).

410

411 Comparison of gut metagenomes in co-infected chickens with and without lauric acid

412 Addition of lauric acid increased the relative abundance of Clostridium sensu stricto 1 and

413 Weissella but decreased the relative amount of Escherichia Shigella in the jejunum compared to

414 the co-infection group without supplementing lauric acid. Nonetheless, no significance was

415 detected in this comparison. In addition, supplementation of lauric acid did not apparently affect

416 the cecal microbiota between these two groups.

37 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

417 Discussion

418 By exploring microbial composition in normal chickens, the major microbial genera in the

419 jejunum were Lactobacillus and Clostridium sensu stricto 1, followed by other unclassified

420 bacteria, Weissella, Enterococcus, Escherichia Shigella, and Staphylococcus. Bacteroides was

421 the most abundant group in the cecum, and the remaining taxa were sequentially other

422 unclassified bacteria, Escherichia Shigella, Eisenbergiella, and Anaerotruncus. Side by side

423 treatments of C. perfringens and Eimeria altered microbial community compositions,

424 significantly in jejunal microbiota. In this study, challenge of CP1 increased the abundance of

425 Clostridium sensu stricto 1, Escherichia Shigella, and Weissella in the jejunum, but significantly

426 decreased the population of Lactobacillus. Infection of Eimeria significantly increased the

427 abundance of Weissella and Staphylococcus, but decreased the amount of Lactobacillus and

428 Clostridium sensu stricto 1. Co-infection with C. perfringens and Eimeria led to significant

429 increment of Clostridium sensu stricto 1, increased abundance of Escherichia Shigella, but

430 decrements of Lactobacillus, Weissella and Staphylococcus. Specifically, it decreases the α-

431 diversity index of the small intestinal microbial community, promoting single dominance of

432 Clostridium sensu stricto 1 reaching the relative abundance to 71.89%. On the other hand, six

433 NE cases shared similar microbial community profile observed in PCA, indicating there exists a

434 certain microbiota contributory to the disease. With more NE severity, higher relative abundance

435 of Clostridium sensu stricto 1 but lower relative amount of Lactobacillus in jejunal microbiota

436 was noted.

437 Several studies has been shown C. perfringens challenge decreased the population of

438 Lactobacillus in ileum [20, 51]. Lactobacilli are known as lactic acid producing bacteria and

439 shown to have protection at intestinal barrier by competition with pathogens. They are also able

38 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

440 to induce immunomodulation and ferment carbohydrates into lactic acids that lower the pH of

441 the intestinal environment to inhibit growth of acid-sensitive pathogenic bacteria [52, 53].

442 Therefore, suppression of lactobacilli is regularly considered beneficial to growth and

443 colonization of enteric pathogen. This study first demonstrated the decrement of lactobacilli in

444 jejunum following challenge of C. perfringens alone and in conjunction with Eimeria. The

445 change of this taxon following the NE severity indicates that decrement of Lactobacillus may

446 play a role in the development of NE. In addition, the increased abundance of Escherichia

447 Shigella was also observed after the challenge of C. perfringens and co-infection with C.

448 perfringens and Eimeria. This genus includes enteric pathogens, which can colonize in the

449 intestines of both humans and chickens, consequently triggering specific diseases [54]. Some

450 studies indicated that the increment of Escherichia Shigella in ileum was correlate with NE [55,

451 56]. Nevertheless, our study found that C. perfringens challenge could increase the abundance of

452 Escherichia Shigella but the increment was not in accordance with NE occurrence. Furthermore,

453 the reduction of this taxa abundance was noticed in lauric acid supplementing group which has

454 higher number of NE cases. Those finding reflected a contradiction for this genus participating in

455 NE development. Last but not least, a reduced abundance of Weissella in the jejunum of NE

456 afflicted chickens was also noted. Another study reported similar result in cecal micorbiota after

457 C. perfringens challenge [18]. Weissella are lactic acid bacteria and belong to the family of the

458 Leuconostocaceae. They harbor probiotic properties and can generate several products with

459 prebiotic potential [57]. It may interact with C. perfringens as other lactic acid bacteria, but its

460 role in NE development is unclear. More studies will be needed to elucidate the relationship

461 between Weissella and NE.

39 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

462 In current study, significant overgrowth of Clostridium sensu stricto 1 was associated with NE

463 and the infection of Eimeria precedent to challenge of C. perfringens exerted synergistic effects

464 on the overrepresentation. This correlation was consistently demonstrated by analyses of

465 metagenomeSeq, STAMP, and LEfSe. The STAMP and LEfSe further showed C. perfringens

466 was significantly overrepresented in NE groups. However, such significance was not identified

467 by metagenomeSeq when C. perfringens was targeted. This result indicates that, in addition to C.

468 perfringens, other bacteria under the same genus of Clostridium sensu stricto 1 also played a role

469 in contributing to the development of disease. The Clostridium genus is well-classified into 19

470 clusters by phylogenetic analysis [58]. Clostridium sensu stricto are grouped around the type

471 species Clostridium butyricum and belong to the Clostridium cluster 1within the

472 family [59]. Clostridium sensu stricto 1 contains C. perfringens and other real Clostridium

473 species. Their members are generally perceived as pathogenic [60] as well as interpreted as an

474 indicator of a less healthy microbiota [61]. This suggestion coincides with our finding that C.

475 perfringens challenge on its own is not capable of causing significant abundance of Clostridium

476 sensu stricto 1 and unable to produce more NE case observed. Future research is recommended

477 to clarify the role of other members of Clostridium sensu stricto 1 in the pathogenesis of NE.

478 Single infection of Eimeria could not produce NE in the present study. The treatment reduced

479 the relative abundance of Clostridium sensu stricto 1 and Lactobacillus, but significantly

480 increased Weissella and Staphylococcus in jejunal microbiota. Eimeria infection has been shown

481 to provide nutrients for C. perfringens to grow and cause physical damage to gut epithelium, thus

482 facilitating the colonization and proliferation of C. perfringens [8, 62, 63]. However, the

483 inoculation of Eimeria into normal chickens did not elicit overgrowth of Clostridium sensu

484 stricto 1 and C. perfringens except challenging with exogenous C. perfringens. In contrast,

40 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

485 challenge of C. perfringens alone and in conjunction with Eimeria both promote proliferation of

486 Clostridium sensu stricto 1 and NE case. This indicates that the amount of commensal C.

487 perfringens in the jejunum under Eimeria infection is not sufficient to reach the significant

488 abundance of Clostridium sensu stricto 1 or C. perfringens, subsequently promoting the

489 occurrence of NE. Therefore, it is reasonable to suggest that the quantity of Clostridium sensu

490 stricto 1 or C. perfringens in jejunum is critical for the onset of proliferation. A recent study used

491 commensal C. perfringens, the isolate from normal chicken, to challenge broiler and reproduce

492 NE in conjunction with infection of E. maxima [64]. This result also highlighted that not the

493 specific C. perfringens strain but the exogenous addition of C. perfringens played the key in

494 achieving the consequence. Accordingly, the methodology to inhibit overgrowth of Clostridium

495 sensu stricto 1 or C. perfringens in small intestines will be the straightforward strategy to prevent

496 NE.

497 Recent studies have been shown that cecal microbiota had a prominent role in feed efficiency [65]

498 and received increasing attention in terms of diseases [66] and metabolism [67]. In this study, the

499 result of PCoA and PCA demonstrated that microbial communities in the jejunum were different

500 from those in the cecum. Side by side treatments of C. perfringens and Eimeria promoted

501 microbial shifts with biological significance in the jejunum but minimal fluctuations in taxa

502 abundance in the cecum. Comparatively, jejunal microbiota was more significant than cecal

503 microbiota to address characteristic gut microbiota contributory to NE by means of

504 metagenomeSeq and LEfSe analysis. The reason might be that cecal microbiota is demonstrated

505 more diverse than other intestinal sections [68] and inhibits higher amounts of microbes (1010-

506 1011 CFU/g) than those in the jejunum (108-109 CFU/g) [69]. Those may provide the buffer

507 effect on microbial changes in cecal microbiota. Besides, preferential colonization of C.

41 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

508 perfringens on mucosa of the small intestine [33] may also contribute to less amount of C.

509 perfringens into cecum, hence adverse to elicit significant changes in cecal microbiota.

510 Medium-chain fatty acids (MCFAs) such as lauric acid are a family of saturated 6- to 12-

511 carbon fatty acids from plants and documented beneficial effects on intestinal health and

512 microbial growth inhibition [70-72]. The mechanism for their bactericidal activity is not fully

513 understood. Relative studies showed that they could act as nonionic surfactants to become

514 incorporated into the bacterial cell membrane, as well as diffuse through cell membranes and

515 create pores, changing membrane permeability and leading to cell death [73-75]. In this work,

516 lauric acid attracted interest due to its inexpensiveness and natural properties, including strong

517 antibacterial effects against C. perfringens and no inhibitory effect on Eimeria infection [76].

518 Based on Timbermont’s study, lauric acid was most effective in inhibiting the growth of C.

519 perfringens strain in vitro. Given a supplementary dose of 0.4 kg/ton in feed caused a significant

520 decrease in NE incidence (from 50% down to 25%) compared with the infected, untreated

521 control group [29]. This study followed the dose and used experimental grade product of lauric

522 acid to evaluate the effects on NE incidence and intestinal microbiota. However, the addition of

523 lauric acid did not reduce the incidence of NE. For intestinal microbiota, lauric acid neither

524 exerted the inhibitory effect against proliferation of C. perfringens nor elevated the level of

525 beneficial bacteria, such as Lactobacillus and Bifidobacterium. But, the relative abundance of

526 Escherichia Shigella was decreased without affecting the incidence. Since lauric acid has

527 different grade of products, such as experimental or food grade, the contradictory result may

528 attribute to the influence of different formula on the absorptive efficiency of this compound.

529 MCFAs are hydrophobic and partly absorbed through the stomach mucosa. Hence, their

42 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

530 triacylglycerols are considered as a desirable formula for feed additive because they can be

531 absorbed intact into intestinal epithelial enterocytes via this form [77].

532 In summary, significant overgrowth of Clostridium sensu stricto 1 in jejunum was recognized

533 as the major microbiota contributory to NE. In addition to C. perfringens, other member within

534 Clostridium sensu stricto 1 was also found to participate in disease development. The decrement

535 of Lactobacillus following the NE severity indicated that lactobacilli also participate in the

536 progress of disease. These taxa showed counteractive effects in their functions as well as in the

537 bacterial abundance, attempting to maintain the homeostasis of jejunal microbiota in chickens.

538 Therefore, manipulations to inhibit multiplication of Clostridium sensu stricto 1 and C.

539 perfringens and to rehabilitate the dominant Lactobacillus population in the jejunum should be

540 the niche for developing effective strategies to prevent NE.

541

542 Acknowledgments

543 The authors would like to thank Dr. John F. Prescott (University of Guelph, Ontario, Canada) for

544 providing netB positive C. perfringens strain CP1. This work was supported by the USDA,

545 National Institute of Food and Agriculture (CRIS Project Accession Number 1014508) and the

546 College of Veterinary Medicine, Mississippi State University.

547

548 References

549 1. Cooper KK, Songer JG, Uzal FA. Diagnosing clostridial enteric disease in poultry. J Vet

550 Diagn Invest. 2013;25(3):314-27. doi: 10.1177/1040638713483468. PubMed PMID:

551 23572451.

43 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

552 2. Martin TG, Smyth JA. Prevalence of netB among some clinical isolates of Clostridium

553 perfringens from animals in the United States. Vet Microbiol. 2009;136(1-2):202-5. Epub

554 2008/12/17. doi: 10.1016/j.vetmic.2008.10.026. PubMed PMID: 19081686.

555 3. Van Der Sluis W. Clostridial enteritis is an often underestimated problem. World poultry.

556 2000;16(7):42-3.

557 4. Wade B, Keyburn. A. The true cost of necrotic enteritis. World Poultry. 2015; 31:16–7.

558 5. Liu D, Guo Y, Wang Z, Yuan J. Exogenous lysozyme influences Clostridium perfringens

559 colonization and intestinal barrier function in broiler chickens. Avian Pathol.

560 2010;39(1):17-24. Epub 2010/04/15. doi: 10.1080/03079450903447404. PubMed PMID:

561 20390532.

562 6. Marshall BM, Levy SB. Food animals and antimicrobials: impacts on human health. Clin

563 Microbiol Rev. 2011;24(4):718-33. doi: 10.1128/CMR.00002-11. PubMed PMID:

564 21976606; PubMed Central PMCID: PMCPMC3194830.

565 7. Van Immerseel F, De Buck J, Pasmans F, Huyghebaert G, Haesebrouck F, Ducatelle R.

566 Clostridium perfringens in poultry: an emerging threat for animal and public health. Avian

567 Pathol. 2004;33(6):537-49. Epub 2005/03/15. doi: 10.1080/03079450400013162. PubMed

568 PMID: 15763720.

569 8. Van Immerseel F, Rood JI, Moore RJ, Titball RW. Rethinking our understanding of the

570 pathogenesis of necrotic enteritis in chickens. Trends Microbiol. 2009;17(1):32-6. Epub

571 2008/11/04. doi: 10.1016/j.tim.2008.09.005. PubMed PMID: 18977143.

572 9. Casewell M, Friis C, Marco E, McMullin P, Phillips I. The European ban on growth-

573 promoting antibiotics and emerging consequences for human and animal health. J

44 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

574 Antimicrob Chemother. 2003;52(2):159-61. Epub 2003/07/03. doi: 10.1093/jac/dkg313.

575 PubMed PMID: 12837737.

576 10. Timbermont L, Haesebrouck F, Ducatelle R, Van Immerseel F. Necrotic enteritis in

577 broilers: an updated review on the pathogenesis. Avian Pathol. 2011;40(4):341-7. Epub

578 2011/08/05. doi: 10.1080/03079457.2011.590967. PubMed PMID: 21812711.

579 11. Gaucher ML, Quessy S, Letellier A, Arsenault J, Boulianne M. Impact of a drug-free

580 program on broiler chicken growth performances, gut health, Clostridium perfringens and

581 Campylobacter jejuni occurrences at the farm level. Poult Sci. 2015;94(8):1791-801. Epub

582 2015/06/07. doi: 10.3382/ps/pev142. PubMed PMID: 26047674.

583 12. Rehman HU, Vahjen W, Awad WA, Zentek J. Indigenous bacteria and bacterial metabolic

584 products in the gastrointestinal tract of broiler chickens. Arch Anim Nutr. 2007;61(5):319-

585 35. Epub 2007/11/23. doi: 10.1080/17450390701556817. PubMed PMID: 18030916.

586 13. Pan D, Yu Z. Intestinal microbiome of poultry and its interaction with host and diet. Gut

587 Microbes. 2014;5(1):108-19. doi: 10.4161/gmic.26945. PubMed PMID: 24256702;

588 PubMed Central PMCID: PMCPMC4049927.

589 14. Yin Y, Lei F, Zhu L, Li S, Wu Z, Zhang R, et al. Exposure of different bacterial inocula to

590 newborn chicken affects gut microbiota development and ileum gene expression. ISME J.

591 2010;4(3):367-76. doi: 10.1038/ismej.2009.128. PubMed PMID: 19956274.

592 15. Mwangi WN, Beal RK, Powers C, Wu X, Humphrey T, Watson M, et al. Regional and

593 global changes in TCRalphabeta T cell repertoires in the gut are dependent upon the

594 complexity of the enteric microflora. Dev Comp Immunol. 2010;34(4):406-17. doi:

595 10.1016/j.dci.2009.11.009. PubMed PMID: 19945480.

45 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

596 16. Crhanova M, Hradecka H, Faldynova M, Matulova M, Havlickova H, Sisak F, et al.

597 Immune response of chicken gut to natural colonization by gut microflora and to

598 Salmonella enterica serovar enteritidis infection. Infect Immun. 2011;79(7):2755-63. doi:

599 10.1128/IAI.01375-10. PubMed PMID: 21555397; PubMed Central PMCID:

600 PMCPMC3191970.

601 17. Feng Y, Gong J, Yu H, Jin Y, Zhu J, Han Y. Identification of changes in the composition

602 of ileal bacterial microbiota of broiler chickens infected with Clostridium perfringens. Vet

603 Microbiol. 2010;140(1-2):116-21. doi: 10.1016/j.vetmic.2009.07.001. PubMed PMID:

604 19647376.

605 18. Stanley D, Keyburn AL, Denman SE, Moore RJ. Changes in the caecal microflora of

606 chickens following Clostridium perfringens challenge to induce necrotic enteritis. Vet

607 Microbiol. 2012;159(1-2):155-62. Epub 2012/04/11. doi: 10.1016/j.vetmic.2012.03.032.

608 PubMed PMID: 22487456.

609 19. Stanley D, Wu SB, Rodgers N, Swick RA, Moore RJ. Differential responses of cecal

610 microbiota to fishmeal, Eimeria and Clostridium perfringens in a necrotic enteritis

611 challenge model in chickens. PLoS One. 2014;9(8):e104739. doi:

612 10.1371/journal.pone.0104739. PubMed PMID: 25167074; PubMed Central PMCID:

613 PMCPMC4148237.

614 20. Li Z, Wang W, Liu D, Guo Y. Effects of Lactobacillus acidophilus on gut microbiota

615 composition in broilers challenged with Clostridium perfringens. PLoS One.

616 2017;12(11):e0188634. doi: 10.1371/journal.pone.0188634. PubMed PMID: 29190649;

617 PubMed Central PMCID: PMCPMC5708699.

46 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

618 21. Dittmar E, Beyer P, Fischer D, Schafer V, Schoepe H, Bauer K, et al. Necrotizing

619 enterocolitis of the neonate with Clostridium perfringens: diagnosis, clinical course, and

620 role of alpha toxin. Eur J Pediatr. 2008;167(8):891-5. doi: 10.1007/s00431-007-0614-9.

621 PubMed PMID: 17952466.

622 22. Heida FH, van Zoonen AG, Hulscher JB, te Kiefte BJ, Wessels R, Kooi EM, et al. A

623 Necrotizing Enterocolitis-Associated Gut Microbiota Is Present in the Meconium: Results

624 of a Prospective Study. Clin Infect Dis. 2016;62(7):863-70. Epub 2016/01/21. doi:

625 10.1093/cid/ciw016. PubMed PMID: 26787171.

626 23. Dahiya JP, Wilkie DC, Van Kessel AG, Drew MD. Potential strategies for controlling

627 necrotic enteritis in broiler chickens in post-antibiotic era. Anim Feed Sci Technol.

628 2006;129(1-2):60-88. doi: 10.1016/j.anifeedsci.2005.12.003.

629 24. Caly DL, D'Inca R, Auclair E, Drider D. Alternatives to Antibiotics to Prevent Necrotic

630 Enteritis in Broiler Chickens: A Microbiologist's Perspective. Front Microbiol.

631 2015;6:1336. doi: 10.3389/fmicb.2015.01336. PubMed PMID: 26648920; PubMed Central

632 PMCID: PMCPMC4664614.

633 25. Hermans D, Martel A, Garmyn A, Verlinden M, Heyndrickx M, Gantois I, et al.

634 Application of medium-chain fatty acids in drinking water increases Campylobacter jejuni

635 colonization threshold in broiler chicks. Poult Sci. 2012;91(7):1733-8. doi:

636 10.3382/ps.2011-02106. PubMed PMID: 22700521.

637 26. Van Immerseel F, De Buck J, Boyen F, Bohez L, Pasmans F, Volf J, et al. Medium-chain

638 fatty acids decrease colonization and invasion through hilA suppression shortly after

639 infection of chickens with Salmonella enterica serovar Enteritidis. Appl Environ

47 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

640 Microbiol. 2004;70(6):3582-7. doi: 10.1128/AEM.70.6.3582-3587.2004. PubMed PMID:

641 15184160; PubMed Central PMCID: PMCPMC427757.

642 27. Zeiger K, Popp J, Becker A, Hankel J, Visscher C, Klein G, et al. Lauric acid as feed

643 additive - An approach to reducing Campylobacter spp. in broiler meat. PLoS One.

644 2017;12(4):e0175693. doi: 10.1371/journal.pone.0175693. PubMed PMID: 28419122;

645 PubMed Central PMCID: PMCPMC5395180.

646 28. Skrivanova E, Marounek M, Dlouha G, Kanka J. Susceptibility of Clostridium perfringens

647 to C-C fatty acids. Lett Appl Microbiol. 2005;41(1):77-81. doi: 10.1111/j.1472-

648 765X.2005.01709.x. PubMed PMID: 15960756.

649 29. Timbermont L, Lanckriet A, Dewulf J, Nollet N, Schwarzer K, Haesebrouck F, et al.

650 Control of Clostridium perfringens-induced necrotic enteritis in broilers by target-released

651 butyric acid, fatty acids and essential oils. Avian Pathol. 2010;39(2):117-21. doi:

652 10.1080/03079451003610586. PubMed PMID: 20390546.

653 30. Wu SB, Stanley D, Rodgers N, Swick RA, Moore RJ. Two necrotic enteritis predisposing

654 factors, dietary fishmeal and Eimeria infection, induce large changes in the caecal

655 microbiota of broiler chickens. Vet Microbiol. 2014;169(3-4):188-97. doi:

656 10.1016/j.vetmic.2014.01.007. PubMed PMID: 24522272.

657 31. Prescott JF, Smyth JA, Shojadoost B, Vince A. Experimental reproduction of necrotic

658 enteritis in chickens: a review. Avian Pathol. 2016;45(3):317-22. doi:

659 10.1080/03079457.2016.1141345. PubMed PMID: 26813025.

660 32. Shojadoost B, Vince AR, Prescott JF. The successful experimental induction of necrotic

661 enteritis in chickens by Clostridium perfringens: a critical review. Vet Res. 2012;43:74.

48 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

662 Epub 2012/10/30. doi: 10.1186/1297-9716-43-74. PubMed PMID: 23101966; PubMed

663 Central PMCID: PMCPMC3546943.

664 33. Prescott JF, Parreira VR, Mehdizadeh Gohari I, Lepp D, Gong J. The pathogenesis of

665 necrotic enteritis in chickens: what we know and what we need to know: a review. Avian

666 Pathol. 2016;45(3):288-94. doi: 10.1080/03079457.2016.1139688. PubMed PMID:

667 26813023.

668 34. Moore RJ. Necrotic enteritis predisposing factors in broiler chickens. Avian Pathol.

669 2016;45(3):275-81. doi: 10.1080/03079457.2016.1150587. PubMed PMID: 26926926.

670 35. Xu S, Lin Y, Zeng D, Zhou M, Zeng Y, Wang H, et al. Bacillus licheniformis normalize

671 the ileum microbiota of chickens infected with necrotic enteritis. Sci Rep. 2018;8(1):1744.

672 doi: 10.1038/s41598-018-20059-z. PubMed PMID: 29379124; PubMed Central PMCID:

673 PMCPMC5789067.

674 36. Lin Y, Xu S, Zeng D, Ni X, Zhou M, Zeng Y, et al. Disruption in the cecal microbiota of

675 chickens challenged with Clostridium perfringens and other factors was alleviated by

676 Bacillus licheniformis supplementation. PLoS One. 2017;12(8):e0182426. doi:

677 10.1371/journal.pone.0182426. PubMed PMID: 28771569; PubMed Central PMCID:

678 PMCPMC5542615.

679 37. Branton SL, Reece FN, Hagler WM, Jr. Influence of a wheat diet on mortality of broiler

680 chickens associated with necrotic enteritis. Poult Sci. 1987;66(8):1326-30. Epub

681 1987/08/01. PubMed PMID: 2891128.

682 38. Thompson DR, Parreira VR, Kulkarni RR, Prescott JF. Live attenuated vaccine-based

683 control of necrotic enteritis of broiler chickens. Vet Microbiol. 2006;113(1-2):25-34. Epub

684 2005/11/18. doi: 10.1016/j.vetmic.2005.10.015. PubMed PMID: 16289639.

49 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

685 39. Jiang Y, Kulkarni RR, Parreira VR, Prescott JF. Immunization of broiler chickens against

686 Clostridium perfringens-induced necrotic enteritis using purified recombinant

687 immunogenic proteins. Avian Dis. 2009;53(3):409-15. Epub 2009/10/24. doi:

688 10.1637/8656-021109-Reg.1. PubMed PMID: 19848081.

689 40. Yu Q, Lepp D, Mehdizadeh Gohari I, Wu T, Zhou H, Yin X, et al. The Agr-like quorum

690 sensing system is required for necrotic enteritis pathogenesis in poultry caused by

691 Clostridium perfringens. Infect Immun. 2017. Epub 2017/04/05. doi: 10.1128/iai.00975-16.

692 PubMed PMID: 28373356.

693 41. Zhou H, Lepp D, Pei Y, Liu M, Yin X, Ma R, et al. Influence of pCP1NetB ancillary genes

694 on the virulence of Clostridium perfringens poultry necrotic enteritis strain CP1. Gut

695 Pathog. 2017;9:6. doi: 10.1186/s13099-016-0152-y. PubMed PMID: 28127404; PubMed

696 Central PMCID: PMCPMC5251324.

697 42. Keyburn AL, Sheedy SA, Ford ME, Williamson MM, Awad MM, Rood JI, et al. Alpha-

698 toxin of Clostridium perfringens is not an essential virulence factor in necrotic enteritis in

699 chickens. Infect Immun. 2006;74(11):6496-500. Epub 2006/08/23. doi: 10.1128/iai.00806-

700 06. PubMed PMID: 16923791; PubMed Central PMCID: PMCPmc1695520.

701 43. Magoc T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome

702 assemblies. Bioinformatics. 2011;27(21):2957-63. doi: 10.1093/bioinformatics/btr507.

703 PubMed PMID: 21903629; PubMed Central PMCID: PMCPMC3198573.

704 44. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al.

705 QIIME allows analysis of high-throughput community sequencing data. Nat Methods.

706 2010;7(5):335-6. doi: 10.1038/nmeth.f.303. PubMed PMID: 20383131; PubMed Central

707 PMCID: PMCPMC3156573.

50 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

708 45. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and

709 speed of chimera detection. Bioinformatics. 2011;27(16):2194-200. doi:

710 10.1093/bioinformatics/btr381. PubMed PMID: 21700674; PubMed Central PMCID:

711 PMCPMC3150044.

712 46. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics.

713 2010;26(19):2460-1. doi: 10.1093/bioinformatics/btq461. PubMed PMID: 20709691.

714 47. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat

715 Methods. 2013;10(10):996-8. Epub 2013/08/21. doi: 10.1038/nmeth.2604. PubMed PMID:

716 23955772.

717 48. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment

718 of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol.

719 2007;73(16):5261-7. doi: 10.1128/AEM.00062-07. PubMed PMID: 17586664; PubMed

720 Central PMCID: PMCPMC1950982.

721 49. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis of taxonomic

722 and functional profiles. Bioinformatics. 2014;30(21):3123-4. doi:

723 10.1093/bioinformatics/btu494. PubMed PMID: 25061070; PubMed Central PMCID:

724 PMCPMC4609014.

725 50. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic

726 biomarker discovery and explanation. Genome Biol. 2011;12(6):R60. doi: 10.1186/gb-

727 2011-12-6-r60. PubMed PMID: 21702898; PubMed Central PMCID: PMCPMC3218848.

728 51. Antonissen G, Eeckhaut V, Van Driessche K, Onrust L, Haesebrouck F, Ducatelle R, et al.

729 Microbial shifts associated with necrotic enteritis. Avian Pathol. 2016;45(3):308-12. doi:

730 10.1080/03079457.2016.1152625. PubMed PMID: 26950294.

51 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

731 52. Sengupta R, Altermann E, Anderson RC, McNabb WC, Moughan PJ, Roy NC. The role of

732 cell surface architecture of lactobacilli in host-microbe interactions in the gastrointestinal

733 tract. Mediators Inflamm. 2013;2013:237921. Epub 2013/04/12. doi:

734 10.1155/2013/237921. PubMed PMID: 23576850; PubMed Central PMCID:

735 PMCPMC3610365.

736 53. Belenguer A, Duncan SH, Holtrop G, Anderson SE, Lobley GE, Flint HJ. Impact of pH on

737 lactate formation and utilization by human fecal microbial communities. Appl Environ

738 Microbiol. 2007;73(20):6526-33. doi: 10.1128/AEM.00508-07. PubMed PMID: 17766450;

739 PubMed Central PMCID: PMCPMC2075063.

740 54. Mora A, Herrera A, Mamani R, Lopez C, Alonso MP, Blanco JE, et al. Recent emergence

741 of clonal group O25b:K1:H4-B2-ST131 ibeA strains among Escherichia coli poultry

742 isolates, including CTX-M-9-producing strains, and comparison with clinical human

743 isolates. Appl Environ Microbiol. 2010;76(21):6991-7. doi: 10.1128/AEM.01112-10.

744 PubMed PMID: 20817805; PubMed Central PMCID: PMCPMC2976257.

745 55. Du E, Gan L, Li Z, Wang W, Liu D, Guo Y. In vitro antibacterial activity of thymol and

746 carvacrol and their effects on broiler chickens challenged with Clostridium perfringens. J

747 Anim Sci Biotechnol. 2015;6:58. doi: 10.1186/s40104-015-0055-7. PubMed PMID:

748 26705471; PubMed Central PMCID: PMCPMC4690362.

749 56. Du E, Wang W, Gan L, Li Z, Guo S, Guo Y. Effects of thymol and carvacrol

750 supplementation on intestinal integrity and immune responses of broiler chickens

751 challenged with Clostridium perfringens. J Anim Sci Biotechnol. 2016;7:19. doi:

752 10.1186/s40104-016-0079-7. PubMed PMID: 27006768; PubMed Central PMCID:

753 PMCPMC4802587.

52 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

754 57. Fusco V, Quero GM, Cho GS, Kabisch J, Meske D, Neve H, et al. The genus Weissella:

755 taxonomy, ecology and biotechnological potential. Front Microbiol. 2015;6:155. doi:

756 10.3389/fmicb.2015.00155. PubMed PMID: 25852652; PubMed Central PMCID:

757 PMCPMC4362408.

758 58. Collins MD, Lawson PA, Willems A, Cordoba JJ, Fernandez-Garayzabal J, Garcia P, et al.

759 The phylogeny of the genus Clostridium: proposal of five new genera and eleven new

760 species combinations. Int J Syst Bacteriol. 1994;44(4):812-26. Epub 1994/10/01. doi:

761 10.1099/00207713-44-4-812. PubMed PMID: 7981107.

762 59. Stackebrandt E, Kramer I, Swiderski J, Hippe H. Phylogenetic basis for a taxonomic

763 dissection of the genus Clostridium. FEMS Immunol Med Microbiol. 1999;24(3):253-8.

764 Epub 1999/07/09. PubMed PMID: 10397308.

765 60. Rajilic-Stojanovic M, de Vos WM. The first 1000 cultured species of the human

766 gastrointestinal microbiota. FEMS Microbiol Rev. 2014;38(5):996-1047. doi:

767 10.1111/1574-6976.12075. PubMed PMID: 24861948; PubMed Central PMCID:

768 PMCPMC4262072.

769 61. Lakshminarayanan B, Harris HM, Coakley M, O'Sullivan O, Stanton C, Pruteanu M, et al.

770 Prevalence and characterization of Clostridium perfringens from the faecal microbiota of

771 elderly Irish subjects. J Med Microbiol. 2013;62(Pt 3):457-66. Epub 2012/12/12. doi:

772 10.1099/jmm.0.052258-0. PubMed PMID: 23222860.

773 62. Williams RB, Marshall RN, La Ragione RM, Catchpole J. A new method for the

774 experimental production of necrotic enteritis and its use for studies on the relationships

775 between necrotic enteritis, coccidiosis and anticoccidial vaccination of chickens. Parasitol

53 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

776 Res. 2003;90(1):19-26. Epub 2003/05/14. doi: 10.1007/s00436-002-0803-4. PubMed

777 PMID: 12743800.

778 63. Williams RB. Intercurrent coccidiosis and necrotic enteritis of chickens: rational,

779 integrated disease management by maintenance of gut integrity. Avian Pathol.

780 2005;34(3):159-80. Epub 2005/09/30. doi: 10.1080/03079450500112195. PubMed PMID:

781 16191699.

782 64. Li C, Lillehoj HS, Gadde UD, Ritter D, Oh S. Characterization of Clostridium perfringens

783 Strains Isolated from Healthy and Necrotic Enteritis-Afflicted Broiler Chickens. Avian

784 Dis. 2017;61(2):178-85. Epub 2017/07/01. doi: 10.1637/11507-093016-Reg.1. PubMed

785 PMID: 28665720.

786 65. Yan W, Sun C, Yuan J, Yang N. Gut metagenomic analysis reveals prominent roles of

787 Lactobacillus and cecal microbiota in chicken feed efficiency. Sci Rep. 2017;7:45308. doi:

788 10.1038/srep45308. PubMed PMID: 28349946.

789 66. Wohlgemuth S, Keller S, Kertscher R, Stadion M, Haller D, Kisling S, et al. Intestinal

790 steroid profiles and microbiota composition in colitic mice. Gut Microbes. 2011;2(3):159-

791 66. doi: 10.4161/gmic.2.3.16104. PubMed PMID: 21869607.

792 67. Stanley D, Denman SE, Hughes RJ, Geier MS, Crowley TM, Chen H, et al. Intestinal

793 microbiota associated with differential feed conversion efficiency in chickens. Appl

794 Microbiol Biotechnol. 2012;96(5):1361-9. Epub 2012/01/18. doi: 10.1007/s00253-011-

795 3847-5. PubMed PMID: 22249719.

796 68. Xiao Y, Xiang Y, Zhou W, Chen J, Li K, Yang H. Microbial community mapping in

797 intestinal tract of broiler chicken. Poult Sci. 2017;96(5):1387-93. doi: 10.3382/ps/pew372.

798 PubMed PMID: 28339527.

54 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

799 69. Yeoman CJ, Chia N, Jeraldo P, Sipos M, Goldenfeld ND, White BA. The microbiome of

800 the chicken gastrointestinal tract. Anim Health Res Rev. 2012;13(1):89-99. doi:

801 10.1017/S1466252312000138. PubMed PMID: 22853945.

802 70. Dierick N, Michiels J, Van Nevel C. Effect of medium chain fatty acids and benzoic acid,

803 as alternatives for antibiotics, on growth and some gut parameters in piglets. Commun

804 Agric Appl Biol Sci. 2004;69(2):187-90. Epub 2004/11/25. PubMed PMID: 15560218.

805 71. Bertevello PL, De Nardi L, Torrinhas RS, Logullo AF, Waitzberg DL. Partial replacement

806 of omega-6 fatty acids with medium-chain triglycerides, but not olive oil, improves colon

807 cytokine response and damage in experimental colitis. JPEN J Parenter Enteral Nutr.

808 2012;36(4):442-8. Epub 2012/01/25. doi: 10.1177/0148607111421788. PubMed PMID:

809 22269895.

810 72. Zentek J, Buchheit-Renko S, Manner K, Pieper R, Vahjen W. Intestinal concentrations of

811 free and encapsulated dietary medium-chain fatty acids and effects on gastric microbial

812 ecology and bacterial metabolic products in the digestive tract of piglets. Arch Anim Nutr.

813 2012;66(1):14-26. Epub 2012/03/09. PubMed PMID: 22397093.

814 73. Desbois AP, Smith VJ. Antibacterial free fatty acids: activities, mechanisms of action and

815 biotechnological potential. Appl Microbiol Biotechnol. 2010;85(6):1629-42. doi:

816 10.1007/s00253-009-2355-3. PubMed PMID: 19956944.

817 74. Altieri C, Bevilacqua A, Cardillo D, Sinigaglia M. Effectiveness of fatty acids and their

818 monoglycerides against gram-negative pathogens. Int J Food Sci Tech. 2009;44(2):359-66.

819 doi: 10.1111/j.1365-2621.2008.01744.x.

55 bioRxiv preprint doi: https://doi.org/10.1101/434449; this version posted October 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

820 75. Bergsson G, Arnfinnsson J, Steingrimsson O, Thormar H. Killing of Gram-positive cocci

821 by fatty acids and monoglycerides. APMIS. 2001;109(10):670-8. Epub 2002/03/14.

822 PubMed PMID: 11890570.

823 76. Mathis G, T. P. van Dam J, Corujo Fernández A, L. Hofacre C. Effect of an organic acids

824 and medium-chain fatty acids containing product in feed on the course of artificial Necrotic

825 Enteritis infection in broiler chickens2018.

826 77. Zentek J, Buchheit-Renko S, Ferrara F, Vahjen W, Van Kessel AG, Pieper R. Nutritional

827 and physiological role of medium-chain triglycerides and medium-chain fatty acids in

828 piglets. Anim Health Res Rev. 2011;12(1):83-93. doi: 10.1017/S1466252311000089.

829 PubMed PMID: 21676342.

830

831 Supporting information

832 Figure S1. Rarefaction curve by groups.

833 Figure S2. Comparison of genera abundance between jejunal and cecal microbiota by STAMP

834 with Welch's t-test.

835 Figure S3. Rank abundance curve by groups.

836 Figure S4. Comparison of genera (A) and species (B) abundance between AJ and EJ, BJ and EJ,

837 and CJ and EJ groups by STAMP with Welch's t-test.

56